Skip to main content
Top
Published in: CNS Drugs 4/2019

01-04-2019 | Opioids | Review Article

Expanding Role of NMDA Receptor Antagonists in the Management of Pain

Authors: Denise Kreutzwiser, Qutaiba A. Tawfic

Published in: CNS Drugs | Issue 4/2019

Login to get access

Abstract

Pain management is complex regardless of whether the pain is acute or chronic in nature or non-cancer or cancer related. In addition, relatively few pain pharmacotherapy options with adequate efficacy and safety data currently exist. Consequently, interest in the role of NMDA receptor antagonists as a pharmacological pain management strategy has surfaced. This narrative review provides an overview of the NMDA receptor and elaborates on the pharmacotherapeutic profile and pain management literature findings for the following NMDA receptor antagonists: ketamine, memantine, dextromethorphan, and magnesium. The literature on this topic is characterized by small studies, many of which exhibit methodological flaws. To date, ketamine is the most studied NMDA receptor antagonist for both acute and chronic pain management. Although further research about NMDA receptor antagonists for analgesia is needed and the optimal dosage/administration regimens for these drugs have yet to be determined, ketamine appears to hold the most promise and may be of particular value in the perioperative pain management realm.
Literature
2.
3.
go back to reference Chou R, Gordon DB, de Leon-Casasola OA, Rosenberg JM, Bickler S, Brennan T, et al. Management of postoperative pain: a clinical practice guideline from the American Pain Society, the American Society of Regional Anesthesia and Pain Medicine, and the American Society of Anesthesiologists’ Committee on Regional Anesthesia, Executive Committee, and Administrative Council. J Pain. 2016;17(2):131–57.CrossRefPubMed Chou R, Gordon DB, de Leon-Casasola OA, Rosenberg JM, Bickler S, Brennan T, et al. Management of postoperative pain: a clinical practice guideline from the American Pain Society, the American Society of Regional Anesthesia and Pain Medicine, and the American Society of Anesthesiologists’ Committee on Regional Anesthesia, Executive Committee, and Administrative Council. J Pain. 2016;17(2):131–57.CrossRefPubMed
4.
go back to reference Schwenk ES, Viscusi ER, Buvanendran A, Hurley RW, Wasan AD, Narouze S, et al. Consensus guidelines on the use of intravenous ketamine infusions for acute pain management from the American Society of Regional Anesthesia and Pain Medicine, the American Academy of Pain Medicine, and the American Society of Anesthesiologists. Reg Anesth Pain Med. 2018;43(5):456–66.PubMedPubMedCentral Schwenk ES, Viscusi ER, Buvanendran A, Hurley RW, Wasan AD, Narouze S, et al. Consensus guidelines on the use of intravenous ketamine infusions for acute pain management from the American Society of Regional Anesthesia and Pain Medicine, the American Academy of Pain Medicine, and the American Society of Anesthesiologists. Reg Anesth Pain Med. 2018;43(5):456–66.PubMedPubMedCentral
5.
go back to reference Gorman AL, Elliott KJ, Inturrisi CE. The d- and l-isomers of methadone bind to the non-competitive site on the N-methyl-D-aspartate (NMDA) receptor in rat forebrain and spinal cord. Neurosci Lett. 1997;223(1):5–8.CrossRefPubMed Gorman AL, Elliott KJ, Inturrisi CE. The d- and l-isomers of methadone bind to the non-competitive site on the N-methyl-D-aspartate (NMDA) receptor in rat forebrain and spinal cord. Neurosci Lett. 1997;223(1):5–8.CrossRefPubMed
6.
go back to reference Ebert B, Andersen S, Krogsgaard-Larsen P. Ketobemidone, methadone and pethidine are non-competitive N-methyl-D-aspartate (NMDA) antagonists in the rat cortex and spinal cord. Neurosci Lett. 1995;187(3):165–8.CrossRefPubMed Ebert B, Andersen S, Krogsgaard-Larsen P. Ketobemidone, methadone and pethidine are non-competitive N-methyl-D-aspartate (NMDA) antagonists in the rat cortex and spinal cord. Neurosci Lett. 1995;187(3):165–8.CrossRefPubMed
7.
go back to reference Davis AM, Inturrisi CE. d-Methadone blocks morphine tolerance and N-methyl-D-aspartate-induced hyperalgesia. J Pharmacol Exp Ther. 1999;289(2):1048–53.PubMed Davis AM, Inturrisi CE. d-Methadone blocks morphine tolerance and N-methyl-D-aspartate-induced hyperalgesia. J Pharmacol Exp Ther. 1999;289(2):1048–53.PubMed
8.
go back to reference Dinis-Oliveira RJ. Metabolomics of methadone: clinical and forensic toxicological implications and variability of dose response. Drug Metab Rev. 2016;48(4):568–76.CrossRefPubMed Dinis-Oliveira RJ. Metabolomics of methadone: clinical and forensic toxicological implications and variability of dose response. Drug Metab Rev. 2016;48(4):568–76.CrossRefPubMed
9.
go back to reference Garrido MJ, Troconiz IF. Methadone: a review of its pharmacokinetic/pharmacodynamic properties. J Pharmacol Toxicol Methods. 1999;42(2):61–6.CrossRefPubMed Garrido MJ, Troconiz IF. Methadone: a review of its pharmacokinetic/pharmacodynamic properties. J Pharmacol Toxicol Methods. 1999;42(2):61–6.CrossRefPubMed
10.
go back to reference Codd EE, Shank RP, Schupsky JJ, Raffa RB. Serotonin and norepinephrine uptake inhibiting activity of centrally acting analgesics: structural determinants and role in antinociception. J Pharmacol Exp Ther. 1995;274(3):1263–70.PubMed Codd EE, Shank RP, Schupsky JJ, Raffa RB. Serotonin and norepinephrine uptake inhibiting activity of centrally acting analgesics: structural determinants and role in antinociception. J Pharmacol Exp Ther. 1995;274(3):1263–70.PubMed
11.
go back to reference Bozic M, Valdivielso JM. The potential of targeting NMDA receptors outside the CNS. Expert Opin Ther Targets. 2015;19(3):399–413.CrossRefPubMed Bozic M, Valdivielso JM. The potential of targeting NMDA receptors outside the CNS. Expert Opin Ther Targets. 2015;19(3):399–413.CrossRefPubMed
12.
go back to reference Paoletti P, Bellone C, Zhou Q. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci. 2013;14(6):383–400.CrossRefPubMed Paoletti P, Bellone C, Zhou Q. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci. 2013;14(6):383–400.CrossRefPubMed
14.
go back to reference Scheetz AJ, Constantine-Paton M. Modulation of NMDA receptor function: implications for vertebrate neural development. FASEB J. 1994;8(10):745–52.CrossRefPubMed Scheetz AJ, Constantine-Paton M. Modulation of NMDA receptor function: implications for vertebrate neural development. FASEB J. 1994;8(10):745–52.CrossRefPubMed
16.
go back to reference Petrenko AB, Yamakura T, Baba H, Shimoji K. The role of N-methyl-D-aspartate (NMDA) receptors in pain: a review. Anesth Analg. 2003;97(4):1108–16.CrossRefPubMed Petrenko AB, Yamakura T, Baba H, Shimoji K. The role of N-methyl-D-aspartate (NMDA) receptors in pain: a review. Anesth Analg. 2003;97(4):1108–16.CrossRefPubMed
17.
18.
go back to reference Woolf CJ. Central sensitization: implications for the diagnosis and treatment of pain. Pain. 2011;152(3 Suppl.):S2–15.CrossRefPubMed Woolf CJ. Central sensitization: implications for the diagnosis and treatment of pain. Pain. 2011;152(3 Suppl.):S2–15.CrossRefPubMed
19.
go back to reference Shanthanna H. Intravenous therapies in the management of neuropathic pain: a review on the use of ketamine and lidocaine in chronic pain management. In: Chukwunonye Udeagha C, editor. neuropathic pain. Rijeka: InTech; 2012. p. 41–78. Shanthanna H. Intravenous therapies in the management of neuropathic pain: a review on the use of ketamine and lidocaine in chronic pain management. In: Chukwunonye Udeagha C, editor. neuropathic pain. Rijeka: InTech; 2012. p. 41–78.
20.
go back to reference Vyklicky V, Korinek M, Smejkalova T, Balik A, Krausova B, Kaniakova M, et al. Structure, function, and pharmacology of NMDA receptor channels. Physiol Res. 2014;63(Suppl. 1):S191–203.PubMed Vyklicky V, Korinek M, Smejkalova T, Balik A, Krausova B, Kaniakova M, et al. Structure, function, and pharmacology of NMDA receptor channels. Physiol Res. 2014;63(Suppl. 1):S191–203.PubMed
21.
go back to reference Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, et al. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev. 2010;62(3):405–96.CrossRefPubMedPubMedCentral Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, et al. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev. 2010;62(3):405–96.CrossRefPubMedPubMedCentral
22.
go back to reference Monyer H, Sprengel R, Schoepfer R, Herb A, Higuchi M, Lomeli H, et al. Heteromeric NMDA receptors: molecular and functional distinction of subtypes. Science. 1992;256(5060):1217–21.CrossRefPubMed Monyer H, Sprengel R, Schoepfer R, Herb A, Higuchi M, Lomeli H, et al. Heteromeric NMDA receptors: molecular and functional distinction of subtypes. Science. 1992;256(5060):1217–21.CrossRefPubMed
24.
go back to reference Lee CH, Lu W, Michel JC, Goehring A, Du J, Song X, et al. NMDA receptor structures reveal subunit arrangement and pore architecture. Nature. 2014;511(7508):191–7.CrossRefPubMedPubMedCentral Lee CH, Lu W, Michel JC, Goehring A, Du J, Song X, et al. NMDA receptor structures reveal subunit arrangement and pore architecture. Nature. 2014;511(7508):191–7.CrossRefPubMedPubMedCentral
26.
27.
go back to reference Pachernegg S, Strutz-Seebohm N, Hollmann M. GluN3 subunit-containing NMDA receptors: not just one-trick ponies. Trends Neurosci. 2012;35(4):240–9.CrossRefPubMed Pachernegg S, Strutz-Seebohm N, Hollmann M. GluN3 subunit-containing NMDA receptors: not just one-trick ponies. Trends Neurosci. 2012;35(4):240–9.CrossRefPubMed
28.
go back to reference Paoletti P. Molecular basis of NMDA receptor functional diversity. Eur J Neurosci. 2011;33(8):1351–65.CrossRefPubMed Paoletti P. Molecular basis of NMDA receptor functional diversity. Eur J Neurosci. 2011;33(8):1351–65.CrossRefPubMed
29.
go back to reference Cull-Candy SG, Leszkiewicz DN. Role of distinct NMDA receptor subtypes at central synapses. Sci STKE. 2004;2004(255):re16. Cull-Candy SG, Leszkiewicz DN. Role of distinct NMDA receptor subtypes at central synapses. Sci STKE. 2004;2004(255):re16.
30.
go back to reference Blanke ML, VanDongen AMJ. Activation mechanisms of the NMDA receptor. In: Van Dongen AM, editor. Biology of the NMDA receptor. Boca Raton: Frontiers in Neuroscience; 2009. Blanke ML, VanDongen AMJ. Activation mechanisms of the NMDA receptor. In: Van Dongen AM, editor. Biology of the NMDA receptor. Boca Raton: Frontiers in Neuroscience; 2009.
31.
go back to reference Strong KL, Jing Y, Prosser AR, Traynelis SF, Liotta DC. NMDA receptor modulators: an updated patent review (2013-2014). Expert Opin Ther Pat. 2014;24(12):1349–66.CrossRefPubMedPubMedCentral Strong KL, Jing Y, Prosser AR, Traynelis SF, Liotta DC. NMDA receptor modulators: an updated patent review (2013-2014). Expert Opin Ther Pat. 2014;24(12):1349–66.CrossRefPubMedPubMedCentral
32.
go back to reference Lipton SA. Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond. Nat Rev Drug Discov. 2006;5(2):160–70.CrossRefPubMed Lipton SA. Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond. Nat Rev Drug Discov. 2006;5(2):160–70.CrossRefPubMed
33.
go back to reference Vadivelu N, Whitney C, Sinatra R. Pain pathways and acute pain processing. In: Sinatra R, De Leon-Cassasola O, Viscusi E, Ginsberg B, editors. Acute pain management. Cambridge: Cambridge University Press; 2009. p. 3–20.CrossRef Vadivelu N, Whitney C, Sinatra R. Pain pathways and acute pain processing. In: Sinatra R, De Leon-Cassasola O, Viscusi E, Ginsberg B, editors. Acute pain management. Cambridge: Cambridge University Press; 2009. p. 3–20.CrossRef
34.
go back to reference World Health Organization. Ketamine update review report. Expert Committee on Drug Dependence Thirty-Seventh Meeting; 2015 Nov 16–20; Geneva. World Health Organization. Ketamine update review report. Expert Committee on Drug Dependence Thirty-Seventh Meeting; 2015 Nov 16–20; Geneva.
35.
go back to reference Peltoniemi MA, Hagelberg NM, Olkkola KT, Saari TI. Ketamine: a review of clinical pharmacokinetics and pharmacodynamics in anesthesia and pain therapy. Clin Pharmacokinet. 2016;55(9):1059–77.CrossRefPubMed Peltoniemi MA, Hagelberg NM, Olkkola KT, Saari TI. Ketamine: a review of clinical pharmacokinetics and pharmacodynamics in anesthesia and pain therapy. Clin Pharmacokinet. 2016;55(9):1059–77.CrossRefPubMed
37.
go back to reference Aroni F, Iacovidou N, Dontas I, Pourzitaki C, Xanthos T. Pharmacological aspects and potential new clinical applications of ketamine: reevaluation of an old drug. J Clin Pharmacol. 2009;49(8):957–64.CrossRefPubMed Aroni F, Iacovidou N, Dontas I, Pourzitaki C, Xanthos T. Pharmacological aspects and potential new clinical applications of ketamine: reevaluation of an old drug. J Clin Pharmacol. 2009;49(8):957–64.CrossRefPubMed
38.
go back to reference Mion G, Villevieille T. Ketamine pharmacology: an update (pharmacodynamics and molecular aspects, recent findings). CNS Neurosci Ther. 2013;19(6):370–80.CrossRefPubMedPubMedCentral Mion G, Villevieille T. Ketamine pharmacology: an update (pharmacodynamics and molecular aspects, recent findings). CNS Neurosci Ther. 2013;19(6):370–80.CrossRefPubMedPubMedCentral
39.
go back to reference Schmid RL, Sandler AN, Katz J. Use and efficacy of low-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes. Pain. 1999;82(2):111–25.CrossRefPubMed Schmid RL, Sandler AN, Katz J. Use and efficacy of low-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes. Pain. 1999;82(2):111–25.CrossRefPubMed
40.
go back to reference Orser BA, Pennefather PS, MacDonald JF. Multiple mechanisms of ketamine blockade of N-methyl-D-aspartate receptors. Anesthesiology. 1997;86(4):903–17.CrossRefPubMed Orser BA, Pennefather PS, MacDonald JF. Multiple mechanisms of ketamine blockade of N-methyl-D-aspartate receptors. Anesthesiology. 1997;86(4):903–17.CrossRefPubMed
41.
go back to reference Mikkelsen S, Ilkjaer S, Brennum J, Borgbjerg FM, Dahl JB. The effect of naloxone on ketamine-induced effects on hyperalgesia and ketamine-induced side effects in humans. Anesthesiology. 1999;90(6):1539–45.CrossRefPubMed Mikkelsen S, Ilkjaer S, Brennum J, Borgbjerg FM, Dahl JB. The effect of naloxone on ketamine-induced effects on hyperalgesia and ketamine-induced side effects in humans. Anesthesiology. 1999;90(6):1539–45.CrossRefPubMed
42.
go back to reference Koizuka S, Obata H, Sasaki M, Saito S, Goto F. Systemic ketamine inhibits hypersensitivity after surgery via descending inhibitory pathways in rats. Can J Anaesth. 2005;52(5):498–505.CrossRefPubMed Koizuka S, Obata H, Sasaki M, Saito S, Goto F. Systemic ketamine inhibits hypersensitivity after surgery via descending inhibitory pathways in rats. Can J Anaesth. 2005;52(5):498–505.CrossRefPubMed
43.
44.
go back to reference Gordh T, Karlsten R, Kristensen J. Intervention with spinal NMDA, adenosine, and NO systems for pain modulation. Ann Med. 1995;27(2):229–34.CrossRefPubMed Gordh T, Karlsten R, Kristensen J. Intervention with spinal NMDA, adenosine, and NO systems for pain modulation. Ann Med. 1995;27(2):229–34.CrossRefPubMed
47.
go back to reference Pai A, Heining M. Ketamine: continuing Education in Anaesthesia. Critical Care & Pain. 2007;7(2):59–63. Pai A, Heining M. Ketamine: continuing Education in Anaesthesia. Critical Care & Pain. 2007;7(2):59–63.
48.
go back to reference Farag E, Argalious M, Tetzlaff JE, Sharma D, editor. Basic sciences in anesthesia. New York: Springer Berlin Heidelberg; 2017. Farag E, Argalious M, Tetzlaff JE, Sharma D, editor. Basic sciences in anesthesia. New York: Springer Berlin Heidelberg; 2017.
49.
go back to reference Calvey TN, Williams NE. Principles and practice of pharmacology for anaesthetists. 5th ed. Malden: Blackwell Publishing; 2008: vii, p. 366. Calvey TN, Williams NE. Principles and practice of pharmacology for anaesthetists. 5th ed. Malden: Blackwell Publishing; 2008: vii, p. 366.
50.
go back to reference Dayton PG, Stiller RL, Cook DR, Perel JM. The binding of ketamine to plasma proteins: emphasis on human plasma. Eur J Clin Pharmacol. 1983;24(6):825–31.CrossRefPubMed Dayton PG, Stiller RL, Cook DR, Perel JM. The binding of ketamine to plasma proteins: emphasis on human plasma. Eur J Clin Pharmacol. 1983;24(6):825–31.CrossRefPubMed
51.
go back to reference Domino EF, Domino SE, Smith RE, Domino LE, Goulet JR, Domino KE, et al. Ketamine kinetics in unmedicated and diazepam-premedicated subjects. Clin Pharmacol Ther. 1984;36(5):645–53.CrossRefPubMed Domino EF, Domino SE, Smith RE, Domino LE, Goulet JR, Domino KE, et al. Ketamine kinetics in unmedicated and diazepam-premedicated subjects. Clin Pharmacol Ther. 1984;36(5):645–53.CrossRefPubMed
52.
go back to reference Ihmsen H, Geisslinger G, Schuttler J. Stereoselective pharmacokinetics of ketamine: R(−)-ketamine inhibits the elimination of S(+)-ketamine. Clin Pharmacol Ther. 2001;70(5):431–8.CrossRefPubMed Ihmsen H, Geisslinger G, Schuttler J. Stereoselective pharmacokinetics of ketamine: R(−)-ketamine inhibits the elimination of S(+)-ketamine. Clin Pharmacol Ther. 2001;70(5):431–8.CrossRefPubMed
53.
go back to reference Sigtermans M, Dahan A, Mooren R, Bauer M, Kest B, Sarton E, et al. S(+)-ketamine effect on experimental pain and cardiac output: a population pharmacokinetic-pharmacodynamic modeling study in healthy volunteers. Anesthesiology. 2009;111(4):892–903.CrossRefPubMed Sigtermans M, Dahan A, Mooren R, Bauer M, Kest B, Sarton E, et al. S(+)-ketamine effect on experimental pain and cardiac output: a population pharmacokinetic-pharmacodynamic modeling study in healthy volunteers. Anesthesiology. 2009;111(4):892–903.CrossRefPubMed
54.
go back to reference Rao LK, Flaker AM, Friedel CC, Kharasch ED. Role of cytochrome P4502B6 polymorphisms in ketamine metabolism and clearance. Anesthesiology. 2016;125(6):1103–12.CrossRefPubMed Rao LK, Flaker AM, Friedel CC, Kharasch ED. Role of cytochrome P4502B6 polymorphisms in ketamine metabolism and clearance. Anesthesiology. 2016;125(6):1103–12.CrossRefPubMed
55.
go back to reference Hijazi Y, Boulieu R. Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2002;30(7):853–8.CrossRefPubMed Hijazi Y, Boulieu R. Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2002;30(7):853–8.CrossRefPubMed
56.
go back to reference Yanagihara Y, Kariya S, Ohtani M, Uchino K, Aoyama T, Yamamura Y, et al. Involvement of CYP2B6 in n-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2001;29(6):887–90.PubMed Yanagihara Y, Kariya S, Ohtani M, Uchino K, Aoyama T, Yamamura Y, et al. Involvement of CYP2B6 in n-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2001;29(6):887–90.PubMed
57.
go back to reference Peltoniemi MA, Saari TI, Hagelberg NM, Reponen P, Turpeinen M, Laine K, et al. Exposure to oral S-ketamine is unaffected by itraconazole but greatly increased by ticlopidine. Clin Pharmacol Ther. 2011;90(2):296–302.CrossRefPubMed Peltoniemi MA, Saari TI, Hagelberg NM, Reponen P, Turpeinen M, Laine K, et al. Exposure to oral S-ketamine is unaffected by itraconazole but greatly increased by ticlopidine. Clin Pharmacol Ther. 2011;90(2):296–302.CrossRefPubMed
58.
go back to reference Peltoniemi MA, Saari TI, Hagelberg NM, Laine K, Kurkinen KJ, Neuvonen PJ, et al. Rifampicin has a profound effect on the pharmacokinetics of oral S-ketamine and less on intravenous S-ketamine. Basic Clin Pharmacol Toxicol. 2012;111(5):325–32.CrossRefPubMed Peltoniemi MA, Saari TI, Hagelberg NM, Laine K, Kurkinen KJ, Neuvonen PJ, et al. Rifampicin has a profound effect on the pharmacokinetics of oral S-ketamine and less on intravenous S-ketamine. Basic Clin Pharmacol Toxicol. 2012;111(5):325–32.CrossRefPubMed
59.
go back to reference Aida S, Yamakura T, Baba H, Taga K, Fukuda S, Shimoji K. Preemptive analgesia by intravenous low-dose ketamine and epidural morphine in gastrectomy: a randomized double-blind study. Anesthesiology. 2000;92(6):1624–30.CrossRefPubMed Aida S, Yamakura T, Baba H, Taga K, Fukuda S, Shimoji K. Preemptive analgesia by intravenous low-dose ketamine and epidural morphine in gastrectomy: a randomized double-blind study. Anesthesiology. 2000;92(6):1624–30.CrossRefPubMed
60.
go back to reference Adriaenssens G, Vermeyen KM, Hoffmann VL, Mertens E, Adriaensen HF. Postoperative analgesia with i.v. patient-controlled morphine: effect of adding ketamine. Br J Anaesth. 1999;83(3):393–6. Adriaenssens G, Vermeyen KM, Hoffmann VL, Mertens E, Adriaensen HF. Postoperative analgesia with i.v. patient-controlled morphine: effect of adding ketamine. Br J Anaesth. 1999;83(3):393–6.
61.
go back to reference Safavi MHA, Nematollahy Z. Pre-incisional analgesia with intravenous or subcutaneous infiltration of ketamine reduces postoperative pain in patients after open cholecystectomy: a randomized, double-blind, placebo-controlled study. Pain Med. 2011;12(9):1418–26.CrossRefPubMed Safavi MHA, Nematollahy Z. Pre-incisional analgesia with intravenous or subcutaneous infiltration of ketamine reduces postoperative pain in patients after open cholecystectomy: a randomized, double-blind, placebo-controlled study. Pain Med. 2011;12(9):1418–26.CrossRefPubMed
62.
63.
go back to reference Parikh B, Maliwad J, Shah VR. Preventive analgesia: effect of small dose of ketamine on morphine requirement after renal surgery. J Anaesthesiol Clin Pharmacol. 2011;27(4):485–8.CrossRefPubMedPubMedCentral Parikh B, Maliwad J, Shah VR. Preventive analgesia: effect of small dose of ketamine on morphine requirement after renal surgery. J Anaesthesiol Clin Pharmacol. 2011;27(4):485–8.CrossRefPubMedPubMedCentral
64.
go back to reference Kararmaz A, Kaya S, Karaman H, Turhanoglu S, Ozyilmaz MA. Intraoperative intravenous ketamine in combination with epidural analgesia: postoperative analgesia after renal surgery. Anesth Analg. 2003;97(4):1092–6.CrossRefPubMed Kararmaz A, Kaya S, Karaman H, Turhanoglu S, Ozyilmaz MA. Intraoperative intravenous ketamine in combination with epidural analgesia: postoperative analgesia after renal surgery. Anesth Analg. 2003;97(4):1092–6.CrossRefPubMed
65.
go back to reference Kwok RF, Lim J, Chan MT, Gin T, Chiu WK. Preoperative ketamine improves postoperative analgesia after gynecologic laparoscopic surgery. Anesth Analg. 2004;98(4):1044–9.CrossRefPubMed Kwok RF, Lim J, Chan MT, Gin T, Chiu WK. Preoperative ketamine improves postoperative analgesia after gynecologic laparoscopic surgery. Anesth Analg. 2004;98(4):1044–9.CrossRefPubMed
66.
go back to reference Lahtinen P, Kokki H, Hakala T, Hynynen M. S(+)-ketamine as an analgesic adjunct reduces opioid consumption after cardiac surgery. Anesth Analg. 2004;99(5):1295–301.CrossRefPubMed Lahtinen P, Kokki H, Hakala T, Hynynen M. S(+)-ketamine as an analgesic adjunct reduces opioid consumption after cardiac surgery. Anesth Analg. 2004;99(5):1295–301.CrossRefPubMed
67.
go back to reference Remerand F, Le Tendre C, Baud A, Couvret C, Pourrat X, Favard L, et al. The early and delayed analgesic effects of ketamine after total hip arthroplasty: a prospective, randomized, controlled, double-blind study. Anesth Analg. 2009;109(6):1963–71.CrossRefPubMed Remerand F, Le Tendre C, Baud A, Couvret C, Pourrat X, Favard L, et al. The early and delayed analgesic effects of ketamine after total hip arthroplasty: a prospective, randomized, controlled, double-blind study. Anesth Analg. 2009;109(6):1963–71.CrossRefPubMed
68.
go back to reference Aveline C, Gautier JF, Vautier P, Cognet F, Hetet HL, Attali JY, et al. Postoperative analgesia and early rehabilitation after total knee replacement: a comparison of continuous low-dose intravenous ketamine versus nefopam. Eur J Pain. 2009;13(6):613–9.CrossRefPubMed Aveline C, Gautier JF, Vautier P, Cognet F, Hetet HL, Attali JY, et al. Postoperative analgesia and early rehabilitation after total knee replacement: a comparison of continuous low-dose intravenous ketamine versus nefopam. Eur J Pain. 2009;13(6):613–9.CrossRefPubMed
69.
go back to reference Yamauchi M, Asano M, Watanabe M, Iwasaki S, Furuse S, Namiki A. Continuous low-dose ketamine improves the analgesic effects of fentanyl patient-controlled analgesia after cervical spine surgery. Anesth Analg. 2008;107(3):1041–4.CrossRefPubMed Yamauchi M, Asano M, Watanabe M, Iwasaki S, Furuse S, Namiki A. Continuous low-dose ketamine improves the analgesic effects of fentanyl patient-controlled analgesia after cervical spine surgery. Anesth Analg. 2008;107(3):1041–4.CrossRefPubMed
70.
go back to reference Loftus RW, Yeager MP, Clark JA, Brown JR, Abdu WA, Sengupta DK, et al. Intraoperative ketamine reduces perioperative opiate consumption in opiate-dependent patients with chronic back pain undergoing back surgery. Anesthesiology. 2010;113(3):639–46.PubMed Loftus RW, Yeager MP, Clark JA, Brown JR, Abdu WA, Sengupta DK, et al. Intraoperative ketamine reduces perioperative opiate consumption in opiate-dependent patients with chronic back pain undergoing back surgery. Anesthesiology. 2010;113(3):639–46.PubMed
71.
go back to reference Subramaniam K, Subramaniam B, Steinbrook RA. Ketamine as adjuvant analgesic to opioids: a quantitative and qualitative systematic review. Anesth Analg. 2004;99(2):482–95.CrossRefPubMed Subramaniam K, Subramaniam B, Steinbrook RA. Ketamine as adjuvant analgesic to opioids: a quantitative and qualitative systematic review. Anesth Analg. 2004;99(2):482–95.CrossRefPubMed
72.
go back to reference Elia N, Tramer MR. Ketamine and postoperative pain: a quantitative systematic review of randomised trials. Pain. 2005;113(1–2):61–70.CrossRefPubMed Elia N, Tramer MR. Ketamine and postoperative pain: a quantitative systematic review of randomised trials. Pain. 2005;113(1–2):61–70.CrossRefPubMed
73.
go back to reference Carstensen M, Moller AM. Adding ketamine to morphine for intravenous patient-controlled analgesia for acute postoperative pain: a qualitative review of randomized trials. Br J Anaesth. 2010;104(4):401–6.CrossRefPubMed Carstensen M, Moller AM. Adding ketamine to morphine for intravenous patient-controlled analgesia for acute postoperative pain: a qualitative review of randomized trials. Br J Anaesth. 2010;104(4):401–6.CrossRefPubMed
74.
go back to reference Laskowski K, Stirling A, McKay WP, Lim HJ. A systematic review of intravenous ketamine for postoperative analgesia. Can J Anaesth. 2011;58(10):911–23.CrossRefPubMed Laskowski K, Stirling A, McKay WP, Lim HJ. A systematic review of intravenous ketamine for postoperative analgesia. Can J Anaesth. 2011;58(10):911–23.CrossRefPubMed
75.
go back to reference Assouline B, Tramer MR, Kreienbuhl L, Elia N. Benefit and harm of adding ketamine to an opioid in a patient-controlled analgesia device for the control of postoperative pain: systematic review and meta-analyses of randomized controlled trials with trial sequential analyses. Pain. 2016;157(12):2854–64.CrossRefPubMed Assouline B, Tramer MR, Kreienbuhl L, Elia N. Benefit and harm of adding ketamine to an opioid in a patient-controlled analgesia device for the control of postoperative pain: systematic review and meta-analyses of randomized controlled trials with trial sequential analyses. Pain. 2016;157(12):2854–64.CrossRefPubMed
76.
go back to reference Wang L, Johnston B, Kaushal A, Cheng D, Zhu F, Martin J. Ketamine added to morphine or hydromorphone patient-controlled analgesia for acute postoperative pain in adults: a systematic review and meta-analysis of randomized trials. Can J Anaesth. 2016;63(3):311–25.CrossRefPubMed Wang L, Johnston B, Kaushal A, Cheng D, Zhu F, Martin J. Ketamine added to morphine or hydromorphone patient-controlled analgesia for acute postoperative pain in adults: a systematic review and meta-analysis of randomized trials. Can J Anaesth. 2016;63(3):311–25.CrossRefPubMed
77.
go back to reference Dahmani S, Michelet D, Abback PS, Wood C, Brasher C, Nivoche Y, et al. Ketamine for perioperative pain management in children: a meta-analysis of published studies. Paediatr Anaesth. 2011;21(6):636–52.CrossRefPubMed Dahmani S, Michelet D, Abback PS, Wood C, Brasher C, Nivoche Y, et al. Ketamine for perioperative pain management in children: a meta-analysis of published studies. Paediatr Anaesth. 2011;21(6):636–52.CrossRefPubMed
78.
go back to reference Pendi A, Field R, Farhan SD, Eichler M, Bederman SS. Perioperative ketamine for analgesia in spine surgery: a meta-analysis of randomized controlled trials. Spine (Phila Pa 1976). 2018;43(5):E299–307. Pendi A, Field R, Farhan SD, Eichler M, Bederman SS. Perioperative ketamine for analgesia in spine surgery: a meta-analysis of randomized controlled trials. Spine (Phila Pa 1976). 2018;43(5):E299–307.
79.
go back to reference Dahi-Taleghani M, Fazli B, Ghasemi M, Vosoughian M, Dabbagh A. Effect of intravenous patient controlled ketamine analgesiaon postoperative pain in opium abusers. Anesth Pain Med. 2014;4(1):e14129.PubMedPubMedCentral Dahi-Taleghani M, Fazli B, Ghasemi M, Vosoughian M, Dabbagh A. Effect of intravenous patient controlled ketamine analgesiaon postoperative pain in opium abusers. Anesth Pain Med. 2014;4(1):e14129.PubMedPubMedCentral
80.
81.
go back to reference Guignard B, Coste C, Costes H, Sessler DI, Lebrault C, Morris W, et al. Supplementing desflurane-remifentanil anesthesia with small-dose ketamine reduces perioperative opioid analgesic requirements. Anesth Analg. 2002;95(1):103–8.CrossRefPubMed Guignard B, Coste C, Costes H, Sessler DI, Lebrault C, Morris W, et al. Supplementing desflurane-remifentanil anesthesia with small-dose ketamine reduces perioperative opioid analgesic requirements. Anesth Analg. 2002;95(1):103–8.CrossRefPubMed
82.
go back to reference Sen H, Sizlan A, Yanarates O, Emirkadi H, Ozkan S, Dagli G, et al. A comparison of gabapentin and ketamine in acute and chronic pain after hysterectomy. Anesth Analg. 2009;109(5):1645–50.CrossRefPubMed Sen H, Sizlan A, Yanarates O, Emirkadi H, Ozkan S, Dagli G, et al. A comparison of gabapentin and ketamine in acute and chronic pain after hysterectomy. Anesth Analg. 2009;109(5):1645–50.CrossRefPubMed
83.
go back to reference McNicol ED, Schumann R, Haroutounian S. A systematic review and meta-analysis of ketamine for the prevention of persistent post-surgical pain. Acta Anaesthesiol Scand. 2014;58(10):1199–213.CrossRefPubMed McNicol ED, Schumann R, Haroutounian S. A systematic review and meta-analysis of ketamine for the prevention of persistent post-surgical pain. Acta Anaesthesiol Scand. 2014;58(10):1199–213.CrossRefPubMed
84.
go back to reference Klatt E, Zumbrunn T, Bandschapp O, Girard T, Ruppen W. Intra- and postoperative intravenous ketamine does not prevent chronic pain: a systematic review and meta-analysis. Scand J Pain. 2015;7(1):42–54.CrossRefPubMed Klatt E, Zumbrunn T, Bandschapp O, Girard T, Ruppen W. Intra- and postoperative intravenous ketamine does not prevent chronic pain: a systematic review and meta-analysis. Scand J Pain. 2015;7(1):42–54.CrossRefPubMed
85.
go back to reference Chaparro LE, Smith SA, Moore RA, Wiffen PJ, Gilron I. Pharmacotherapy for the prevention of chronic pain after surgery in adults. Cochrane Database Syst Rev. 2013;(7):CD008307. Chaparro LE, Smith SA, Moore RA, Wiffen PJ, Gilron I. Pharmacotherapy for the prevention of chronic pain after surgery in adults. Cochrane Database Syst Rev. 2013;(7):CD008307.
86.
go back to reference Cohen SP, Bhatia A, Buvanendran A, Schwenk ES, Wasan AD, Hurley RW, et al. Consensus guidelines on the use of intravenous ketamine infusions for chronic pain from the American Society of Regional Anesthesia and Pain Medicine, the American Academy of Pain Medicine, and the American Society of Anesthesiologists. Reg Anesth Pain Med. 2018;43(5):521–46.PubMedPubMedCentral Cohen SP, Bhatia A, Buvanendran A, Schwenk ES, Wasan AD, Hurley RW, et al. Consensus guidelines on the use of intravenous ketamine infusions for chronic pain from the American Society of Regional Anesthesia and Pain Medicine, the American Academy of Pain Medicine, and the American Society of Anesthesiologists. Reg Anesth Pain Med. 2018;43(5):521–46.PubMedPubMedCentral
87.
go back to reference Blonk MI, Koder BG, van den Bemt PM, Huygen FJ. Use of oral ketamine in chronic pain management: a review. Eur J Pain. 2010;14(5):466–72.CrossRefPubMed Blonk MI, Koder BG, van den Bemt PM, Huygen FJ. Use of oral ketamine in chronic pain management: a review. Eur J Pain. 2010;14(5):466–72.CrossRefPubMed
88.
go back to reference Marchetti F, Coutaux A, Bellanger A, Magneux C, Bourgeois P, Mion G. Efficacy and safety of oral ketamine for the relief of intractable chronic pain: a retrospective 5-year study of 51 patients. Eur J Pain. 2015;19(7):984–93.CrossRefPubMed Marchetti F, Coutaux A, Bellanger A, Magneux C, Bourgeois P, Mion G. Efficacy and safety of oral ketamine for the relief of intractable chronic pain: a retrospective 5-year study of 51 patients. Eur J Pain. 2015;19(7):984–93.CrossRefPubMed
89.
go back to reference Rabi J. Topical ketamine: a review of the history, mechanisms, uses, safety, and future Int J Pharm Compd. 2016;20(2):107–13. Rabi J. Topical ketamine: a review of the history, mechanisms, uses, safety, and future Int J Pharm Compd. 2016;20(2):107–13.
90.
go back to reference Kopsky DJ, Keppel Hesselink JM, Bhaskar A, Hariton G, Romanenko V, Casale R. Analgesic effects of topical ketamine. Minerva Anestesiol. 2015;81(4):440–9.PubMed Kopsky DJ, Keppel Hesselink JM, Bhaskar A, Hariton G, Romanenko V, Casale R. Analgesic effects of topical ketamine. Minerva Anestesiol. 2015;81(4):440–9.PubMed
91.
go back to reference Finch PM, Knudsen L, Drummond PD. Reduction of allodynia in patients with complex regional pain syndrome: a double-blind placebo-controlled trial of topical ketamine. Pain. 2009;146(1–2):18–25.CrossRefPubMed Finch PM, Knudsen L, Drummond PD. Reduction of allodynia in patients with complex regional pain syndrome: a double-blind placebo-controlled trial of topical ketamine. Pain. 2009;146(1–2):18–25.CrossRefPubMed
92.
go back to reference Rabi J, Minori J, Abad H, Lee R, Gittler M. Topical ketamine 10% for neuropathic pain in spinal cord injury patients: an open-label trial. Int J Pharm Compd. 2016;20(6):517–20.PubMed Rabi J, Minori J, Abad H, Lee R, Gittler M. Topical ketamine 10% for neuropathic pain in spinal cord injury patients: an open-label trial. Int J Pharm Compd. 2016;20(6):517–20.PubMed
94.
go back to reference Jonkman K, van de Donk T, Dahan A. Ketamine for cancer pain: what is the evidence? Curr Opin Support Palliat Care. 2017;11(2):88–92.CrossRefPubMed Jonkman K, van de Donk T, Dahan A. Ketamine for cancer pain: what is the evidence? Curr Opin Support Palliat Care. 2017;11(2):88–92.CrossRefPubMed
95.
go back to reference Chung WJ, Pharo GH. Successful use of ketamine infusion in the treatment of intractable cancer pain in an outpatient. J Pain Symptom Manage. 2007;33(1):2–5.CrossRefPubMed Chung WJ, Pharo GH. Successful use of ketamine infusion in the treatment of intractable cancer pain in an outpatient. J Pain Symptom Manage. 2007;33(1):2–5.CrossRefPubMed
96.
go back to reference Ripamonti CI, Santini D, Maranzano E, Berti M, Roila F, Group EGW. Management of cancer pain: ESMO clinical practice guidelines. Ann Oncol. 2012;23(Suppl. 7):vii139–54. Ripamonti CI, Santini D, Maranzano E, Berti M, Roila F, Group EGW. Management of cancer pain: ESMO clinical practice guidelines. Ann Oncol. 2012;23(Suppl. 7):vii139–54.
97.
go back to reference Mercadante S, Arcuri E, Tirelli W, Casuccio A. Analgesic effect of intravenous ketamine in cancer patients on morphine therapy: a randomized, controlled, double-blind, crossover, double-dose study. J Pain Symptom Manage. 2000;20(4):246–52.CrossRefPubMed Mercadante S, Arcuri E, Tirelli W, Casuccio A. Analgesic effect of intravenous ketamine in cancer patients on morphine therapy: a randomized, controlled, double-blind, crossover, double-dose study. J Pain Symptom Manage. 2000;20(4):246–52.CrossRefPubMed
98.
go back to reference Salas S, Frasca M, Planchet-Barraud B, Burucoa B, Pascal M, Lapiana JM, et al. Ketamine analgesic effect by continuous intravenous infusion in refractory cancer pain: considerations about the clinical research in palliative care. J Palliat Med. 2012;15(3):287–93.CrossRefPubMed Salas S, Frasca M, Planchet-Barraud B, Burucoa B, Pascal M, Lapiana JM, et al. Ketamine analgesic effect by continuous intravenous infusion in refractory cancer pain: considerations about the clinical research in palliative care. J Palliat Med. 2012;15(3):287–93.CrossRefPubMed
99.
go back to reference Bell RF, Eccleston C, Kalso EA. Ketamine as an adjuvant to opioids for cancer pain. Cochrane Database Syst Rev. 2017;6:CD003351. Bell RF, Eccleston C, Kalso EA. Ketamine as an adjuvant to opioids for cancer pain. Cochrane Database Syst Rev. 2017;6:CD003351.
100.
go back to reference Bai X, Yan Y, Canfield S, Muravyeva MY, Kikuchi C, Zaja I, et al. Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated mitochondrial pathway. Anesth Analg. 2013;116(4):869–80.CrossRefPubMedPubMedCentral Bai X, Yan Y, Canfield S, Muravyeva MY, Kikuchi C, Zaja I, et al. Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated mitochondrial pathway. Anesth Analg. 2013;116(4):869–80.CrossRefPubMedPubMedCentral
101.
go back to reference Curran HV, Monaghan L. In and out of the K-hole: a comparison of the acute and residual effects of ketamine in frequent and infrequent ketamine users. Addiction. 2001;96(5):749–60.CrossRefPubMed Curran HV, Monaghan L. In and out of the K-hole: a comparison of the acute and residual effects of ketamine in frequent and infrequent ketamine users. Addiction. 2001;96(5):749–60.CrossRefPubMed
102.
go back to reference Hayashi H, Dikkes P, Soriano SG. Repeated administration of ketamine may lead to neuronal degeneration in the developing rat brain. Paediatr Anaesth. 2002;12(9):770–4.CrossRefPubMed Hayashi H, Dikkes P, Soriano SG. Repeated administration of ketamine may lead to neuronal degeneration in the developing rat brain. Paediatr Anaesth. 2002;12(9):770–4.CrossRefPubMed
103.
go back to reference Soriano SG, Liu Q, Li J, Liu JR, Han XH, Kanter JL, et al. Ketamine activates cell cycle signaling and apoptosis in the neonatal rat brain. Anesthesiology. 2010;112(5):1155–63.CrossRefPubMed Soriano SG, Liu Q, Li J, Liu JR, Han XH, Kanter JL, et al. Ketamine activates cell cycle signaling and apoptosis in the neonatal rat brain. Anesthesiology. 2010;112(5):1155–63.CrossRefPubMed
104.
go back to reference Proescholdt M, Heimann A, Kempski O. Neuroprotection of S(+) ketamine isomer in global forebrain ischemia. Brain Res. 2001;904(2):245–51.CrossRefPubMed Proescholdt M, Heimann A, Kempski O. Neuroprotection of S(+) ketamine isomer in global forebrain ischemia. Brain Res. 2001;904(2):245–51.CrossRefPubMed
105.
go back to reference Yan J, Li YR, Zhang Y, Lu Y, Jiang H. Repeated exposure to anesthetic ketamine can negatively impact neurodevelopment in infants: a prospective preliminary clinical study. J Child Neurol. 2014;29(10):1333–8.CrossRefPubMed Yan J, Li YR, Zhang Y, Lu Y, Jiang H. Repeated exposure to anesthetic ketamine can negatively impact neurodevelopment in infants: a prospective preliminary clinical study. J Child Neurol. 2014;29(10):1333–8.CrossRefPubMed
106.
go back to reference Morgan CJ, Curran HV. Acute and chronic effects of ketamine upon human memory: a review. Psychopharmacology. 2006;188(4):408–24.CrossRefPubMed Morgan CJ, Curran HV. Acute and chronic effects of ketamine upon human memory: a review. Psychopharmacology. 2006;188(4):408–24.CrossRefPubMed
107.
go back to reference Chu PS, Ma WK, Wong SC, Chu RW, Cheng CH, Wong S, et al. The destruction of the lower urinary tract by ketamine abuse: a new syndrome? BJU Int. 2008;102(11):1616–22.CrossRefPubMed Chu PS, Ma WK, Wong SC, Chu RW, Cheng CH, Wong S, et al. The destruction of the lower urinary tract by ketamine abuse: a new syndrome? BJU Int. 2008;102(11):1616–22.CrossRefPubMed
108.
go back to reference Sear JW. Ketamine hepato-toxicity in chronic pain management: another example of unexpected toxicity or a predicted result from previous clinical and pre-clinical data? Pain. 2011;152(9):1946–7.CrossRefPubMed Sear JW. Ketamine hepato-toxicity in chronic pain management: another example of unexpected toxicity or a predicted result from previous clinical and pre-clinical data? Pain. 2011;152(9):1946–7.CrossRefPubMed
109.
go back to reference Witt A, Macdonald N, Kirkpatrick P. Memantine hydrochloride. Nat Rev Drug Discov. 2004;3(2):109–10.CrossRefPubMed Witt A, Macdonald N, Kirkpatrick P. Memantine hydrochloride. Nat Rev Drug Discov. 2004;3(2):109–10.CrossRefPubMed
110.
go back to reference Ebixa (memantine hydrochloride) [product monograph]. Montreal (QC): Lundbeck Canada Inc.; Last updated 14 May 2015. Ebixa (memantine hydrochloride) [product monograph]. Montreal (QC): Lundbeck Canada Inc.; Last updated 14 May 2015.
111.
go back to reference Johnson JW, Kotermanski SE. Mechanism of action of memantine. Curr Opin Pharmacol. 2006;6(1):61–7.CrossRefPubMed Johnson JW, Kotermanski SE. Mechanism of action of memantine. Curr Opin Pharmacol. 2006;6(1):61–7.CrossRefPubMed
112.
go back to reference Kavirajan H. Memantine: a comprehensive review of safety and efficacy. Expert Opin Drug Saf. 2009;8(1):89–109.CrossRefPubMed Kavirajan H. Memantine: a comprehensive review of safety and efficacy. Expert Opin Drug Saf. 2009;8(1):89–109.CrossRefPubMed
113.
go back to reference Kornhuber J, Quack G, Danysz W, Jellinger K, Danielczyk W, Gsell W, et al. Therapeutic brain concentration of the NMDA receptor antagonist amantadine. Neuropharmacology. 1995;34(7):713–21.CrossRefPubMed Kornhuber J, Quack G, Danysz W, Jellinger K, Danielczyk W, Gsell W, et al. Therapeutic brain concentration of the NMDA receptor antagonist amantadine. Neuropharmacology. 1995;34(7):713–21.CrossRefPubMed
114.
go back to reference Morel V, Etienne M, Wattiez AS, Dupuis A, Privat AM, Chalus M, et al. Memantine, a promising drug for the prevention of neuropathic pain in rat. Eur J Pharmacol. 2013;721(1–3):382–90.CrossRefPubMed Morel V, Etienne M, Wattiez AS, Dupuis A, Privat AM, Chalus M, et al. Memantine, a promising drug for the prevention of neuropathic pain in rat. Eur J Pharmacol. 2013;721(1–3):382–90.CrossRefPubMed
115.
go back to reference Rahimzadeh P, Imani F, Nikoubakht N, Koleini Z, Faiz SHR, Sayarifard A. A comparative study on the efficacy of oral memantine and placebo for acute postoperative pain in patients undergoing dacryocystorhinostomy (DCR). Anesth Pain Med. 2017;7(3):e45297.CrossRefPubMedPubMedCentral Rahimzadeh P, Imani F, Nikoubakht N, Koleini Z, Faiz SHR, Sayarifard A. A comparative study on the efficacy of oral memantine and placebo for acute postoperative pain in patients undergoing dacryocystorhinostomy (DCR). Anesth Pain Med. 2017;7(3):e45297.CrossRefPubMedPubMedCentral
116.
go back to reference Morel V, Joly D, Villatte C, Dubray C, Durando X, Daulhac L, et al. Memantine before mastectomy prevents post-surgery pain: a randomized, blinded clinical trial in surgical patients. PLoS One. 2016;11(4):e0152741.CrossRefPubMedPubMedCentral Morel V, Joly D, Villatte C, Dubray C, Durando X, Daulhac L, et al. Memantine before mastectomy prevents post-surgery pain: a randomized, blinded clinical trial in surgical patients. PLoS One. 2016;11(4):e0152741.CrossRefPubMedPubMedCentral
117.
go back to reference Schley M, Topfner S, Wiech K, Schaller HE, Konrad CJ, Schmelz M, et al. Continuous brachial plexus blockade in combination with the NMDA receptor antagonist memantine prevents phantom pain in acute traumatic upper limb amputees. Eur J Pain. 2007;11(3):299–308.CrossRefPubMed Schley M, Topfner S, Wiech K, Schaller HE, Konrad CJ, Schmelz M, et al. Continuous brachial plexus blockade in combination with the NMDA receptor antagonist memantine prevents phantom pain in acute traumatic upper limb amputees. Eur J Pain. 2007;11(3):299–308.CrossRefPubMed
118.
go back to reference Eisenberg E, Kleiser A, Dortort A, Haim T, Yarnitsky D. The NMDA (N-methyl-D-aspartate) receptor antagonist memantine in the treatment of postherpetic neuralgia: a double-blind, placebo-controlled study. Eur J Pain. 1998;2(4):321–7.CrossRefPubMed Eisenberg E, Kleiser A, Dortort A, Haim T, Yarnitsky D. The NMDA (N-methyl-D-aspartate) receptor antagonist memantine in the treatment of postherpetic neuralgia: a double-blind, placebo-controlled study. Eur J Pain. 1998;2(4):321–7.CrossRefPubMed
119.
go back to reference Fayed N, Olivan-Blazquez B, Herrera-Mercadal P, Puebla-Guedea M, Perez-Yus MC, Andres E, et al. Changes in metabolites after treatment with memantine in fibromyalgia: a double-blind randomized controlled trial with magnetic resonance spectroscopy with a 6-month follow-up. CNS Neurosci Ther. 2014;20(11):999–1007.CrossRefPubMedPubMedCentral Fayed N, Olivan-Blazquez B, Herrera-Mercadal P, Puebla-Guedea M, Perez-Yus MC, Andres E, et al. Changes in metabolites after treatment with memantine in fibromyalgia: a double-blind randomized controlled trial with magnetic resonance spectroscopy with a 6-month follow-up. CNS Neurosci Ther. 2014;20(11):999–1007.CrossRefPubMedPubMedCentral
120.
go back to reference Maier C, Dertwinkel R, Mansourian N, Hosbach I, Schwenkreis P, Senne I, et al. Efficacy of the NMDA-receptor antagonist memantine in patients with chronic phantom limb pain: results of a randomized double-blinded, placebo-controlled trial. Pain. 2003;103(3):277–83.CrossRefPubMed Maier C, Dertwinkel R, Mansourian N, Hosbach I, Schwenkreis P, Senne I, et al. Efficacy of the NMDA-receptor antagonist memantine in patients with chronic phantom limb pain: results of a randomized double-blinded, placebo-controlled trial. Pain. 2003;103(3):277–83.CrossRefPubMed
121.
go back to reference Nikolajsen L, Gottrup H, Kristensen AG, Jensen TS. Memantine (a N-methyl-D-aspartate receptor antagonist) in the treatment of neuropathic pain after amputation or surgery: a randomized, double-blinded, cross-over study. Anesth Analg. 2000;91(4):960–6.CrossRefPubMed Nikolajsen L, Gottrup H, Kristensen AG, Jensen TS. Memantine (a N-methyl-D-aspartate receptor antagonist) in the treatment of neuropathic pain after amputation or surgery: a randomized, double-blinded, cross-over study. Anesth Analg. 2000;91(4):960–6.CrossRefPubMed
122.
go back to reference Olivan-Blazquez B, Herrera-Mercadal P, Puebla-Guedea M, Perez-Yus MC, Andres E, Fayed N, et al. Efficacy of memantine in the treatment of fibromyalgia: a double-blind, randomised, controlled trial with 6-month follow-up. Pain. 2014;155(12):2517–25.CrossRefPubMed Olivan-Blazquez B, Herrera-Mercadal P, Puebla-Guedea M, Perez-Yus MC, Andres E, Fayed N, et al. Efficacy of memantine in the treatment of fibromyalgia: a double-blind, randomised, controlled trial with 6-month follow-up. Pain. 2014;155(12):2517–25.CrossRefPubMed
123.
go back to reference Sang CN, Booher S, Gilron I, Parada S, Max MB. Dextromethorphan and memantine in painful diabetic neuropathy and postherpetic neuralgia: efficacy and dose-response trials. Anesthesiology. 2002;96(5):1053–61.CrossRefPubMed Sang CN, Booher S, Gilron I, Parada S, Max MB. Dextromethorphan and memantine in painful diabetic neuropathy and postherpetic neuralgia: efficacy and dose-response trials. Anesthesiology. 2002;96(5):1053–61.CrossRefPubMed
124.
go back to reference Schifitto G, Yiannoutsos CT, Simpson DM, Marra CM, Singer EJ, Kolson DL, et al. A placebo-controlled study of memantine for the treatment of human immunodeficiency virus-associated sensory neuropathy. J Neurovirol. 2006;12(4):328–31.CrossRefPubMed Schifitto G, Yiannoutsos CT, Simpson DM, Marra CM, Singer EJ, Kolson DL, et al. A placebo-controlled study of memantine for the treatment of human immunodeficiency virus-associated sensory neuropathy. J Neurovirol. 2006;12(4):328–31.CrossRefPubMed
125.
go back to reference Schwenkreis P, Maier C, Pleger B, Mansourian N, Dertwinkel R, Malin JP, et al. NMDA-mediated mechanisms in cortical excitability changes after limb amputation. Acta Neurol Scand. 2003;108(3):179–84.CrossRefPubMed Schwenkreis P, Maier C, Pleger B, Mansourian N, Dertwinkel R, Malin JP, et al. NMDA-mediated mechanisms in cortical excitability changes after limb amputation. Acta Neurol Scand. 2003;108(3):179–84.CrossRefPubMed
126.
go back to reference Wiech K, Kiefer RT, Topfner S, Preissl H, Braun C, Unertl K, et al. A placebo-controlled randomized crossover trial of the N-methyl-d-aspartic acid receptor antagonist, memantine, in patients with chronic phantom limb pain. Anesth Analg. 2004;98(2):408–13.CrossRefPubMed Wiech K, Kiefer RT, Topfner S, Preissl H, Braun C, Unertl K, et al. A placebo-controlled randomized crossover trial of the N-methyl-d-aspartic acid receptor antagonist, memantine, in patients with chronic phantom limb pain. Anesth Analg. 2004;98(2):408–13.CrossRefPubMed
127.
go back to reference Sinis N, Birbaumer N, Gustin S, Schwarz A, Bredanger S, Becker ST, et al. Memantine treatment of complex regional pain syndrome: a preliminary report of six cases. Clin J Pain. 2007;23(3):237–43.CrossRefPubMed Sinis N, Birbaumer N, Gustin S, Schwarz A, Bredanger S, Becker ST, et al. Memantine treatment of complex regional pain syndrome: a preliminary report of six cases. Clin J Pain. 2007;23(3):237–43.CrossRefPubMed
128.
go back to reference Ahmad-Sabry MH, Shareghi G. Effects of memantine on pain in patients with complex regional pain syndrome: a retrospective study. Middle East J Anaesthesiol. 2015;23(1):51–4.PubMed Ahmad-Sabry MH, Shareghi G. Effects of memantine on pain in patients with complex regional pain syndrome: a retrospective study. Middle East J Anaesthesiol. 2015;23(1):51–4.PubMed
129.
go back to reference Loy BM, Britt RB, Brown JN. Memantine for the treatment of phantom limb pain: a systematic review. J Pain Palliat Care Pharmacother. 2016;30(4):276–83.CrossRefPubMed Loy BM, Britt RB, Brown JN. Memantine for the treatment of phantom limb pain: a systematic review. J Pain Palliat Care Pharmacother. 2016;30(4):276–83.CrossRefPubMed
130.
go back to reference Alviar MJ, Hale T, Dungca M. Pharmacologic interventions for treating phantom limb pain. Cochrane Database Syst Rev. 2016;10:CD006380. Alviar MJ, Hale T, Dungca M. Pharmacologic interventions for treating phantom limb pain. Cochrane Database Syst Rev. 2016;10:CD006380.
131.
go back to reference Pickering G, Morel V. Memantine for the treatment of general neuropathic pain: a narrative review. Fundam Clin Pharmacol. 2018;32(1):4–13.CrossRefPubMed Pickering G, Morel V. Memantine for the treatment of general neuropathic pain: a narrative review. Fundam Clin Pharmacol. 2018;32(1):4–13.CrossRefPubMed
132.
go back to reference Pickering G, Pereira B, Morel V, Tiberghien F, Martin E, Marcaillou F, et al. Rationale and design of a multicenter randomized clinical trial with memantine and dextromethorphan in ketamine-responder patients. Contemp Clin Trials. 2014;38(2):314–20.CrossRefPubMed Pickering G, Pereira B, Morel V, Tiberghien F, Martin E, Marcaillou F, et al. Rationale and design of a multicenter randomized clinical trial with memantine and dextromethorphan in ketamine-responder patients. Contemp Clin Trials. 2014;38(2):314–20.CrossRefPubMed
133.
go back to reference Assarzadegan F, Sistanizad M. Tolerability and efficacy of memantine as add on therapy in patients with migraine. Iran J Pharm Res. 2017;16(2):791–7.PubMedPubMedCentral Assarzadegan F, Sistanizad M. Tolerability and efficacy of memantine as add on therapy in patients with migraine. Iran J Pharm Res. 2017;16(2):791–7.PubMedPubMedCentral
134.
go back to reference Bigal M, Rapoport A, Sheftell F, Tepper D, Tepper S. Memantine in the preventive treatment of refractory migraine. Headache. 2008;48(9):1337–42.CrossRefPubMed Bigal M, Rapoport A, Sheftell F, Tepper D, Tepper S. Memantine in the preventive treatment of refractory migraine. Headache. 2008;48(9):1337–42.CrossRefPubMed
135.
go back to reference Lindelof K, Bendtsen L. Memantine for prophylaxis of chronic tension-type headache: a double-blind, randomized, crossover clinical trial. Cephalalgia. 2009;29(3):314–21.CrossRefPubMed Lindelof K, Bendtsen L. Memantine for prophylaxis of chronic tension-type headache: a double-blind, randomized, crossover clinical trial. Cephalalgia. 2009;29(3):314–21.CrossRefPubMed
136.
go back to reference Noruzzadeh R, Modabbernia A, Aghamollaii V, Ghaffarpour M, Harirchian MH, Salahi S, et al. Memantine for prophylactic treatment of migraine without aura: a randomized double-blind placebo-controlled study. Headache. 2016;56(1):95–103.CrossRefPubMed Noruzzadeh R, Modabbernia A, Aghamollaii V, Ghaffarpour M, Harirchian MH, Salahi S, et al. Memantine for prophylactic treatment of migraine without aura: a randomized double-blind placebo-controlled study. Headache. 2016;56(1):95–103.CrossRefPubMed
137.
go back to reference Nguyen L, Thomas KL, Lucke-Wold BP, Cavendish JZ, Crowe MS, Matsumoto RR. Dextromethorphan: an update on its utility for neurological and neuropsychiatric disorders. Pharmacol Ther. 2016;159:1–22.CrossRefPubMed Nguyen L, Thomas KL, Lucke-Wold BP, Cavendish JZ, Crowe MS, Matsumoto RR. Dextromethorphan: an update on its utility for neurological and neuropsychiatric disorders. Pharmacol Ther. 2016;159:1–22.CrossRefPubMed
138.
go back to reference Gudin J, Fudin J, Nalamachu S. Levorphanol use: past, present and future. Postgrad Med. 2016;128(1):46–53.CrossRefPubMed Gudin J, Fudin J, Nalamachu S. Levorphanol use: past, present and future. Postgrad Med. 2016;128(1):46–53.CrossRefPubMed
139.
go back to reference Taylor CP, Traynelis SF, Siffert J, Pope LE, Matsumoto RR. Pharmacology of dextromethorphan: relevance to dextromethorphan/quinidine (Nuedexta®) clinical use. Pharmacol Ther. 2016;164:170–82.CrossRefPubMed Taylor CP, Traynelis SF, Siffert J, Pope LE, Matsumoto RR. Pharmacology of dextromethorphan: relevance to dextromethorphan/quinidine (Nuedexta®) clinical use. Pharmacol Ther. 2016;164:170–82.CrossRefPubMed
140.
go back to reference Church J, Lodge D, Berry SC. Differential effects of dextrorphan and levorphanol on the excitation of rat spinal neurons by amino acids. Eur J Pharmacol. 1985;111(2):185–90.CrossRefPubMed Church J, Lodge D, Berry SC. Differential effects of dextrorphan and levorphanol on the excitation of rat spinal neurons by amino acids. Eur J Pharmacol. 1985;111(2):185–90.CrossRefPubMed
141.
go back to reference Church J, Jones MG, Davies SN, Lodge D. Antitussive agents as N-methylaspartate antagonists: further studies. Can J Physiol Pharmacol. 1989;67(6):561–7.CrossRefPubMed Church J, Jones MG, Davies SN, Lodge D. Antitussive agents as N-methylaspartate antagonists: further studies. Can J Physiol Pharmacol. 1989;67(6):561–7.CrossRefPubMed
142.
go back to reference Franklin PH, Murray TF. High affinity [3H]dextrorphan binding in rat brain is localized to a noncompetitive antagonist site of the activated N-methyl-D-aspartate receptor-cation channel. Mol Pharmacol. 1992;41(1):134–46.PubMed Franklin PH, Murray TF. High affinity [3H]dextrorphan binding in rat brain is localized to a noncompetitive antagonist site of the activated N-methyl-D-aspartate receptor-cation channel. Mol Pharmacol. 1992;41(1):134–46.PubMed
143.
go back to reference Netzer R, Pflimlin P, Trube G. Dextromethorphan blocks N-methyl-D-aspartate-induced currents and voltage-operated inward currents in cultured cortical neurons. Eur J Pharmacol. 1993;238(2–3):209–16.CrossRefPubMed Netzer R, Pflimlin P, Trube G. Dextromethorphan blocks N-methyl-D-aspartate-induced currents and voltage-operated inward currents in cultured cortical neurons. Eur J Pharmacol. 1993;238(2–3):209–16.CrossRefPubMed
144.
go back to reference Palmer GC. Neuroprotection by NMDA receptor antagonists in a variety of neuropathologies. Curr Drug Targets. 2001;2(3):241–71.CrossRefPubMed Palmer GC. Neuroprotection by NMDA receptor antagonists in a variety of neuropathologies. Curr Drug Targets. 2001;2(3):241–71.CrossRefPubMed
145.
go back to reference Nuedexta (dextromethorphan hydrobromide and quinidine sulfate) capsules [prescribing information]. Aliso Viejo (CA): Avanir Pharmaceuticals, Inc.; last updated Oct 2010. Nuedexta (dextromethorphan hydrobromide and quinidine sulfate) capsules [prescribing information]. Aliso Viejo (CA): Avanir Pharmaceuticals, Inc.; last updated Oct 2010.
146.
go back to reference Miller A, Pratt H, Schiffer RB. Pseudobulbar affect: the spectrum of clinical presentations, etiologies and treatments. Expert Rev Neurother. 2011;11(7):1077–88.CrossRefPubMed Miller A, Pratt H, Schiffer RB. Pseudobulbar affect: the spectrum of clinical presentations, etiologies and treatments. Expert Rev Neurother. 2011;11(7):1077–88.CrossRefPubMed
147.
go back to reference Hollander D, Pradas J, Kaplan R, McLeod HL, Evans WE, Munsat TL. High-dose dextromethorphan in amyotrophic lateral sclerosis: phase I safety and pharmacokinetic studies. Ann Neurol. 1994;36(6):920–4.CrossRefPubMed Hollander D, Pradas J, Kaplan R, McLeod HL, Evans WE, Munsat TL. High-dose dextromethorphan in amyotrophic lateral sclerosis: phase I safety and pharmacokinetic studies. Ann Neurol. 1994;36(6):920–4.CrossRefPubMed
148.
go back to reference Siu A, Drachtman R. Dextromethorphan: a review of N-methyl-d-aspartate receptor antagonist in the management of pain. CNS Drug Rev. 2007;13(1):96–106.CrossRefPubMedPubMedCentral Siu A, Drachtman R. Dextromethorphan: a review of N-methyl-d-aspartate receptor antagonist in the management of pain. CNS Drug Rev. 2007;13(1):96–106.CrossRefPubMedPubMedCentral
149.
go back to reference Pender ES, Parks BR. Toxicity with dextromethorphan-containing preparations: a literature review and report of two additional cases. Pediatr Emerg Care. 1991;7(3):163–5.CrossRefPubMed Pender ES, Parks BR. Toxicity with dextromethorphan-containing preparations: a literature review and report of two additional cases. Pediatr Emerg Care. 1991;7(3):163–5.CrossRefPubMed
150.
go back to reference Duedahl TH, Romsing J, Moiniche S, Dahl JB. A qualitative systematic review of peri-operative dextromethorphan in post-operative pain. Acta Anaesthesiol Scand. 2006;50(1):1–13.CrossRefPubMed Duedahl TH, Romsing J, Moiniche S, Dahl JB. A qualitative systematic review of peri-operative dextromethorphan in post-operative pain. Acta Anaesthesiol Scand. 2006;50(1):1–13.CrossRefPubMed
151.
go back to reference Aiyer R, Mehta N, Gungor S, Gulati A. A systematic review of NMDA receptor antagonists for treatment of neuropathic pain in clinical practice. Clin J Pain. 2018;34(5):450–67.PubMed Aiyer R, Mehta N, Gungor S, Gulati A. A systematic review of NMDA receptor antagonists for treatment of neuropathic pain in clinical practice. Clin J Pain. 2018;34(5):450–67.PubMed
152.
go back to reference Yu A, Haining RL. Comparative contribution to dextromethorphan metabolism by cytochrome P450 isoforms in vitro: can dextromethorphan be used as a dual probe for both CYP2D6 and CYP3A activities? Drug Metab Dispos. 2001;29(11):1514–20.PubMed Yu A, Haining RL. Comparative contribution to dextromethorphan metabolism by cytochrome P450 isoforms in vitro: can dextromethorphan be used as a dual probe for both CYP2D6 and CYP3A activities? Drug Metab Dispos. 2001;29(11):1514–20.PubMed
153.
go back to reference Shin EJ, Bach JH, Lee SY, Kim JM, Lee J, Hong JS, et al. Neuropsychotoxic and neuroprotective potentials of dextromethorphan and its analogs. J Pharmacol Sci. 2011;116(2):137–48.CrossRefPubMed Shin EJ, Bach JH, Lee SY, Kim JM, Lee J, Hong JS, et al. Neuropsychotoxic and neuroprotective potentials of dextromethorphan and its analogs. J Pharmacol Sci. 2011;116(2):137–48.CrossRefPubMed
154.
go back to reference Wu D, Otton SV, Kalow W, Sellers EM. Effects of route of administration on dextromethorphan pharmacokinetics and behavioral response in the rat. J Pharmacol Exp Ther. 1995;274(3):1431–7.PubMed Wu D, Otton SV, Kalow W, Sellers EM. Effects of route of administration on dextromethorphan pharmacokinetics and behavioral response in the rat. J Pharmacol Exp Ther. 1995;274(3):1431–7.PubMed
155.
go back to reference Capon DA, Bochner F, Kerry N, Mikus G, Danz C, Somogyi AA. The influence of CYP2D6 polymorphism and quinidine on the disposition and antitussive effect of dextromethorphan in humans. Clin Pharmacol Ther. 1996;60(3):295–307.CrossRefPubMed Capon DA, Bochner F, Kerry N, Mikus G, Danz C, Somogyi AA. The influence of CYP2D6 polymorphism and quinidine on the disposition and antitussive effect of dextromethorphan in humans. Clin Pharmacol Ther. 1996;60(3):295–307.CrossRefPubMed
156.
go back to reference King MR, Ladha KS, Gelineau AM, Anderson TA. Perioperative dextromethorphan as an adjunct for postoperative pain: a meta-analysis of randomized controlled trials. Anesthesiology. 2016;124(3):696–705.CrossRefPubMed King MR, Ladha KS, Gelineau AM, Anderson TA. Perioperative dextromethorphan as an adjunct for postoperative pain: a meta-analysis of randomized controlled trials. Anesthesiology. 2016;124(3):696–705.CrossRefPubMed
157.
go back to reference Nosotti M, Rosso L, Tosi D, Palleschi A, Mendogni P, Righi I, et al. Preventive analgesia in thoracic surgery: controlled, randomized, double-blinded study. Eur J Cardiothorac Surg. 2015;48(3):428–33 (discussion 34). Nosotti M, Rosso L, Tosi D, Palleschi A, Mendogni P, Righi I, et al. Preventive analgesia in thoracic surgery: controlled, randomized, double-blinded study. Eur J Cardiothorac Surg. 2015;48(3):428–33 (discussion 34).
158.
go back to reference Carlsson KC, Hoem NO, Moberg ER, Mathisen LC. Analgesic effect of dextromethorphan in neuropathic pain. Acta Anaesthesiol Scand. 2004;48(3):328–36.CrossRefPubMed Carlsson KC, Hoem NO, Moberg ER, Mathisen LC. Analgesic effect of dextromethorphan in neuropathic pain. Acta Anaesthesiol Scand. 2004;48(3):328–36.CrossRefPubMed
159.
go back to reference Dudgeon DJ, Bruera E, Gagnon B, Watanabe SM, Allan SJ, Warr DG, et al. A phase III randomized, double-blind, placebo-controlled study evaluating dextromethorphan plus slow-release morphine for chronic cancer pain relief in terminally ill patients. J Pain Symptom Manage. 2007;33(4):365–71.CrossRefPubMed Dudgeon DJ, Bruera E, Gagnon B, Watanabe SM, Allan SJ, Warr DG, et al. A phase III randomized, double-blind, placebo-controlled study evaluating dextromethorphan plus slow-release morphine for chronic cancer pain relief in terminally ill patients. J Pain Symptom Manage. 2007;33(4):365–71.CrossRefPubMed
160.
go back to reference Galer BS, Lee D, Ma T, Nagle B, Schlagheck TG. MorphiDex (morphine sulfate/dextromethorphan hydrobromide combination) in the treatment of chronic pain: three multicenter, randomized, double-blind, controlled clinical trials fail to demonstrate enhanced opioid analgesia or reduction in tolerance. Pain. 2005;115(3):284–95.CrossRefPubMed Galer BS, Lee D, Ma T, Nagle B, Schlagheck TG. MorphiDex (morphine sulfate/dextromethorphan hydrobromide combination) in the treatment of chronic pain: three multicenter, randomized, double-blind, controlled clinical trials fail to demonstrate enhanced opioid analgesia or reduction in tolerance. Pain. 2005;115(3):284–95.CrossRefPubMed
161.
go back to reference Gilron I, Booher SL, Rowan MS, Smoller MS, Max MB. A randomized, controlled trial of high-dose dextromethorphan in facial neuralgias. Neurology. 2000;55(7):964–71.CrossRefPubMed Gilron I, Booher SL, Rowan MS, Smoller MS, Max MB. A randomized, controlled trial of high-dose dextromethorphan in facial neuralgias. Neurology. 2000;55(7):964–71.CrossRefPubMed
162.
go back to reference Heiskanen T, Hartel B, Dahl ML, Seppala T, Kalso E. Analgesic effects of dextromethorphan and morphine in patients with chronic pain. Pain. 2002;96(3):261–7.CrossRefPubMed Heiskanen T, Hartel B, Dahl ML, Seppala T, Kalso E. Analgesic effects of dextromethorphan and morphine in patients with chronic pain. Pain. 2002;96(3):261–7.CrossRefPubMed
163.
go back to reference Katz NP. MorphiDex (MS:DM) double-blind, multiple-dose studies in chronic pain patients. J Pain Symptom Manage. 2000;19(1 Suppl.):S37–41.CrossRefPubMed Katz NP. MorphiDex (MS:DM) double-blind, multiple-dose studies in chronic pain patients. J Pain Symptom Manage. 2000;19(1 Suppl.):S37–41.CrossRefPubMed
164.
go back to reference McQuay HJ, Carroll D, Jadad AR, Glynn CJ, Jack T, Moore RA, et al. Dextromethorphan for the treatment of neuropathic pain: a double-blind randomised controlled crossover trial with integral n-of-1 design. Pain. 1994;59(1):127–33.CrossRefPubMed McQuay HJ, Carroll D, Jadad AR, Glynn CJ, Jack T, Moore RA, et al. Dextromethorphan for the treatment of neuropathic pain: a double-blind randomised controlled crossover trial with integral n-of-1 design. Pain. 1994;59(1):127–33.CrossRefPubMed
165.
go back to reference Mercadante S, Casuccio A, Genovese G. Ineffectiveness of dextromethorphan in cancer pain. J Pain Symptom Manage. 1998;16(5):317–22.CrossRefPubMed Mercadante S, Casuccio A, Genovese G. Ineffectiveness of dextromethorphan in cancer pain. J Pain Symptom Manage. 1998;16(5):317–22.CrossRefPubMed
166.
go back to reference Nelson KA, Park KM, Robinovitz E, Tsigos C, Max MB. High-dose oral dextromethorphan versus placebo in painful diabetic neuropathy and postherpetic neuralgia. Neurology. 1997;48(5):1212–8.CrossRefPubMed Nelson KA, Park KM, Robinovitz E, Tsigos C, Max MB. High-dose oral dextromethorphan versus placebo in painful diabetic neuropathy and postherpetic neuralgia. Neurology. 1997;48(5):1212–8.CrossRefPubMed
167.
go back to reference Srebro D, Vuckovic S, Milovanovic A, Kosutic J, Vujovic KS, Prostran M. Magnesium in pain research: state of the art. Curr Med Chem. 2016 Dec 12. (Epub ahead of print). Srebro D, Vuckovic S, Milovanovic A, Kosutic J, Vujovic KS, Prostran M. Magnesium in pain research: state of the art. Curr Med Chem. 2016 Dec 12. (Epub ahead of print).
168.
go back to reference Yousef AA, Al-deeb AE. A double-blinded randomised controlled study of the value of sequential intravenous and oral magnesium therapy in patients with chronic low back pain with a neuropathic component. Anaesthesia. 2013;68(3):260–6.CrossRefPubMed Yousef AA, Al-deeb AE. A double-blinded randomised controlled study of the value of sequential intravenous and oral magnesium therapy in patients with chronic low back pain with a neuropathic component. Anaesthesia. 2013;68(3):260–6.CrossRefPubMed
169.
go back to reference Bujalska-Zadrozny M, Tatarkiewicz J, Kulik K, Filip M, Naruszewicz M. Magnesium enhances opioid-induced analgesia: what we have learnt in the past decades? Eur J Pharm Sci. 2017;99:113–27.CrossRefPubMed Bujalska-Zadrozny M, Tatarkiewicz J, Kulik K, Filip M, Naruszewicz M. Magnesium enhances opioid-induced analgesia: what we have learnt in the past decades? Eur J Pharm Sci. 2017;99:113–27.CrossRefPubMed
170.
go back to reference Dube L, Granry JC. The therapeutic use of magnesium in anesthesiology, intensive care and emergency medicine: a review. Can J Anaesth. 2003;50(7):732–46.CrossRefPubMed Dube L, Granry JC. The therapeutic use of magnesium in anesthesiology, intensive care and emergency medicine: a review. Can J Anaesth. 2003;50(7):732–46.CrossRefPubMed
171.
go back to reference De Oliveira GS, Jr Castro-Alves LJ, Khan JH, McCarthy RJ. Perioperative systemic magnesium to minimize postoperative pain: a meta-analysis of randomized controlled trials. Anesthesiology. 2013;119(1):178–90.CrossRefPubMed De Oliveira GS, Jr Castro-Alves LJ, Khan JH, McCarthy RJ. Perioperative systemic magnesium to minimize postoperative pain: a meta-analysis of randomized controlled trials. Anesthesiology. 2013;119(1):178–90.CrossRefPubMed
172.
go back to reference Albrecht E, Kirkham KR, Liu SS, Brull R. Peri-operative intravenous administration of magnesium sulphate and postoperative pain: a meta-analysis. Anaesthesia. 2013;68(1):79–90.CrossRefPubMed Albrecht E, Kirkham KR, Liu SS, Brull R. Peri-operative intravenous administration of magnesium sulphate and postoperative pain: a meta-analysis. Anaesthesia. 2013;68(1):79–90.CrossRefPubMed
173.
go back to reference Murphy JD, Paskaradevan J, Eisler LL, Ouanes JP, Tomas VA, Freck EA, et al. Analgesic efficacy of continuous intravenous magnesium infusion as an adjuvant to morphine for postoperative analgesia: a systematic review and meta-analysis. Middle East J Anaesthesiol. 2013;22(1):11–20.PubMed Murphy JD, Paskaradevan J, Eisler LL, Ouanes JP, Tomas VA, Freck EA, et al. Analgesic efficacy of continuous intravenous magnesium infusion as an adjuvant to morphine for postoperative analgesia: a systematic review and meta-analysis. Middle East J Anaesthesiol. 2013;22(1):11–20.PubMed
174.
go back to reference Sousa AM, Rosado GM, Neto Jde S, Guimaraes GM, Ashmawi HA. Magnesium sulfate improves postoperative analgesia in laparoscopic gynecologic surgeries: a double-blind randomized controlled trial. J Clin Anesth. 2016;34:379–84.CrossRefPubMed Sousa AM, Rosado GM, Neto Jde S, Guimaraes GM, Ashmawi HA. Magnesium sulfate improves postoperative analgesia in laparoscopic gynecologic surgeries: a double-blind randomized controlled trial. J Clin Anesth. 2016;34:379–84.CrossRefPubMed
175.
go back to reference Choi H, Parmar N. The use of intravenous magnesium sulphate for acute migraine: meta-analysis of randomized controlled trials. Eur J Emerg Med. 2014;21(1):2–9.PubMed Choi H, Parmar N. The use of intravenous magnesium sulphate for acute migraine: meta-analysis of randomized controlled trials. Eur J Emerg Med. 2014;21(1):2–9.PubMed
176.
go back to reference Chiu HY, Yeh TH, Huang YC, Chen PY. Effects of intravenous and oral magnesium on reducing migraine: a meta-analysis of randomized controlled trials. Pain Physician. 2016;19(1):E97–112.PubMed Chiu HY, Yeh TH, Huang YC, Chen PY. Effects of intravenous and oral magnesium on reducing migraine: a meta-analysis of randomized controlled trials. Pain Physician. 2016;19(1):E97–112.PubMed
177.
go back to reference Fischer SG, Collins S, Boogaard S, Loer SA, Zuurmond WW, Perez RS. Intravenous magnesium for chronic complex regional pain syndrome type 1 (CRPS-1). Pain Med. 2013;14(9):1388–99.CrossRefPubMed Fischer SG, Collins S, Boogaard S, Loer SA, Zuurmond WW, Perez RS. Intravenous magnesium for chronic complex regional pain syndrome type 1 (CRPS-1). Pain Med. 2013;14(9):1388–99.CrossRefPubMed
178.
go back to reference van der Plas AA, Schilder JC, Marinus J, van Hilten JJ. An explanatory study evaluating the muscle relaxant effects of intramuscular magnesium sulphate for dystonia in complex regional pain syndrome. J Pain. 2013;14(11):1341–8.CrossRefPubMed van der Plas AA, Schilder JC, Marinus J, van Hilten JJ. An explanatory study evaluating the muscle relaxant effects of intramuscular magnesium sulphate for dystonia in complex regional pain syndrome. J Pain. 2013;14(11):1341–8.CrossRefPubMed
179.
go back to reference Pickering G, Morel V, Simen E, Cardot JM, Moustafa F, Delage N, et al. Oral magnesium treatment in patients with neuropathic pain: a randomized clinical trial. Magnes Res. 2011;24(2):28–35.PubMed Pickering G, Morel V, Simen E, Cardot JM, Moustafa F, Delage N, et al. Oral magnesium treatment in patients with neuropathic pain: a randomized clinical trial. Magnes Res. 2011;24(2):28–35.PubMed
180.
go back to reference Brill S, Sedgwick PM, Hamann W, Di Vadi PP. Efficacy of intravenous magnesium in neuropathic pain. Br J Anaesth. 2002;89(5):711–4. Brill S, Sedgwick PM, Hamann W, Di Vadi PP. Efficacy of intravenous magnesium in neuropathic pain. Br J Anaesth. 2002;89(5):711–4.
181.
go back to reference Felsby S, Nielsen J, Arendt-Nielsen L, Jensen TS. NMDA receptor blockade in chronic neuropathic pain: a comparison of ketamine and magnesium chloride. Pain. 1996;64(2):283–91.CrossRefPubMed Felsby S, Nielsen J, Arendt-Nielsen L, Jensen TS. NMDA receptor blockade in chronic neuropathic pain: a comparison of ketamine and magnesium chloride. Pain. 1996;64(2):283–91.CrossRefPubMed
182.
go back to reference Kim YH, Lee PB, Oh TK. Is magnesium sulfate effective for pain in chronic postherpetic neuralgia patients comparing with ketamine infusion therapy? J Clin Anesth. 2015;27(4):296–300.CrossRefPubMed Kim YH, Lee PB, Oh TK. Is magnesium sulfate effective for pain in chronic postherpetic neuralgia patients comparing with ketamine infusion therapy? J Clin Anesth. 2015;27(4):296–300.CrossRefPubMed
Metadata
Title
Expanding Role of NMDA Receptor Antagonists in the Management of Pain
Authors
Denise Kreutzwiser
Qutaiba A. Tawfic
Publication date
01-04-2019
Publisher
Springer International Publishing
Published in
CNS Drugs / Issue 4/2019
Print ISSN: 1172-7047
Electronic ISSN: 1179-1934
DOI
https://doi.org/10.1007/s40263-019-00618-2

Other articles of this Issue 4/2019

CNS Drugs 4/2019 Go to the issue